Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
Add more filters

Publication year range
1.
PLoS Pathog ; 19(9): e1011647, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37738244

ABSTRACT

The bacterial microbiota promotes the life cycle of the intestine-dwelling whipworm Trichuris by mediating hatching of parasite eggs ingested by the mammalian host. Despite the enormous disease burden associated with Trichuris colonization, the mechanisms underlying this transkingdom interaction have been obscure. Here, we used a multiscale microscopy approach to define the structural events associated with bacteria-mediated hatching of eggs for the murine model parasite Trichuris muris. Through the combination of scanning electron microscopy (SEM) and serial block face SEM (SBFSEM), we visualized the outer surface morphology of the shell and generated 3D structures of the egg and larva during the hatching process. These images revealed that exposure to hatching-inducing bacteria catalyzed asymmetric degradation of the polar plugs prior to exit by the larva. Unrelated bacteria induced similar loss of electron density and dissolution of the structural integrity of the plugs. Egg hatching was most efficient when high densities of bacteria were bound to the poles. Consistent with the ability of taxonomically distant bacteria to induce hatching, additional results suggest chitinase released from larva within the eggs degrade the plugs from the inside instead of enzymes produced by bacteria in the external environment. These findings define at ultrastructure resolution the evolutionary adaptation of a parasite for the microbe-rich environment of the mammalian gut.


Subject(s)
Microbiota , Trichuris , Mice , Animals , Microscopy, Electron, Scanning , Bacteria , Larva , Ovum , Mammals
2.
Eur J Clin Microbiol Infect Dis ; 43(3): 587-596, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38261158

ABSTRACT

BACKGROUND: Over a billion people are infected with Toxocara canis or T. cati, the roundworms of dogs and cats. Historically, T. canis has been considered the main species responsible for human toxocarosis, but as serodiagnosis cannot discriminate between the two species, this remains unresolved. We used pigs as a relevant large animal model for human infection to assess the migratory pattern of T. cati and T. canis. METHODS: Pigs were inoculated with T. cati or T. canis eggs or PBS (negative controls) and necropsied 14 or 31 days later. Different organs and tissues were examined for parasites and pathological changes. RESULTS: Overall, the two parasite species had a similar migration pattern reaching multiple organs and tissues, including the mesenteric lymph nodes, liver, lungs, and diaphragm. We recovered larvae of both species in the brain, suggesting that T. cati also can cause neurological toxocarosis in humans. Both species induced systemic eosinophilia and histopathological changes in the lungs, livers, and mesenteric lymph nodes. CONCLUSION: This study emphasises the importance of T. cati as a zoonotic agent and the need to develop diagnostic methods that can differentiate between sources of infection in humans.


Subject(s)
Toxocara canis , Toxocariasis , Animals , Humans , Swine , Toxocara , Toxocariasis/diagnosis , Toxocariasis/parasitology , Toxocariasis/pathology
3.
Cell Commun Signal ; 21(1): 297, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37864211

ABSTRACT

BACKGROUND: E. coli O83 (Colinfant Newborn) is a Gram-negative (G-) probiotic bacterium used in the clinic. When administered orally, it reduces allergic sensitisation but not allergic asthma. Intranasal administration offers a non-invasive and convenient delivery method. This route bypasses the gastrointestinal tract and provides direct access to the airways, which are the target of asthma prevention. G- bacteria such as E. coli O83 release outer membrane vesicles (OMVs) to communicate with the environment. Here we investigate whether intranasally administered E. coli O83 OMVs (EcO83-OMVs) can reduce allergic airway inflammation in mice. METHODS: EcO83-OMVs were isolated by ultracentrifugation and characterised their number, morphology (shape and size), composition (proteins and lipopolysaccharide; LPS), recognition by innate receptors (using transfected HEK293 cells) and immunomodulatory potential (in naïve splenocytes and bone marrow-derived dendritic cells; BMDCs). Their allergy-preventive effect was investigated in a mouse model of ovalbumin-induced allergic airway inflammation. RESULTS: EcO83-OMVs are spherical nanoparticles with a size of about 110 nm. They contain LPS and protein cargo. We identified a total of 1120 proteins, 136 of which were enriched in OMVs compared to parent bacteria. Proteins from the flagellum dominated. OMVs activated the pattern recognition receptors TLR2/4/5 as well as NOD1 and NOD2. EcO83-OMVs induced the production of pro- and anti-inflammatory cytokines in splenocytes and BMDCs. Intranasal administration of EcO83-OMVs inhibited airway hyperresponsiveness, and decreased airway eosinophilia, Th2 cytokine production and mucus secretion. CONCLUSIONS: We demonstrate for the first time that intranasally administered OMVs from probiotic G- bacteria have an anti-allergic effect. Our study highlights the advantages of OMVs as a safe platform for the prophylactic treatment of allergy. Video Abstract.


Subject(s)
Asthma , Extracellular Vesicles , Hypersensitivity , Probiotics , Humans , Animals , Mice , Escherichia coli , Lipopolysaccharides , HEK293 Cells , Hypersensitivity/prevention & control , Hypersensitivity/metabolism , Immunity, Innate , Asthma/metabolism , Inflammation/metabolism , Extracellular Vesicles/metabolism , Probiotics/pharmacology
4.
Cytokine ; 156: 155919, 2022 08.
Article in English | MEDLINE | ID: mdl-35649322

ABSTRACT

Regulation of macrophage (Mɸ) function can maintain tissue homeostasis and control inflammation. Parasitic worms (helminths) are potent modulators of host immune and inflammatory responses. They have evolved various strategies to promote immunosuppression, including redirecting phagocytic cells toward a regulatory phenotype. Although soluble products from the whipworm Trichuris suis (TSPs) have shown significant effects on Mɸ function, the mechanisms underlying these modulatory effects are still not well understood. In this study, we find that TSPs suppressed inflammatory cytokines (TNF and IL-6) in Mɸs stimulated with a broad panel of TLR agonists, whilst inducing IL-10. Moreover, M1 markers such as MHCII, CD86, iNOS, and TNF were downregulated in TSP-treated Mɸs, without polarizing them towards an M2-like phenotype. We showed that TSPs could establish a suppressed activation state of Mɸs lasting at least for 72 h, indicating an anti-inflammatory innate training. Moreover, we found that TSPs, via repression of intracellular TNF generation, decreased its secretion rather than interfering with the release of surface-bound TNF. Metabolic analysis showed that TSPs promote oxidative phosphorylation (OXPHOS) without affecting glycolytic rate. Collectively, these findings expand our knowledge on helminth-induced immune modulation and support future investigations into the anti-inflammatory properties of TSPs for therapeutic purposes.


Subject(s)
Trichuriasis , Trichuris , Animals , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Cytokines/metabolism , Macrophages/metabolism , Trichuriasis/metabolism , Trichuriasis/parasitology , Trichuris/metabolism
5.
Lupus ; 31(4): 415-423, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35202548

ABSTRACT

AIM: Current treatment of Systemic Lupus Erythematosus (SLE) is suboptimal and causes broad immunosuppression. Therapeutic use of helminths or helminth products has been suggested for autoimmune diseases such as SLE. In the present study, we evaluated possible immunomodulating effects of adult body fluid (ABF) from Ascaris suum on peripheral blood mononuclear cells (PBMCs) from SLE patients in an ex vivo setup. METHODS: PBMCs from SLE patients and healthy controls (HC) were isolated and stimulated ex vivo with ABF and Toll-like receptor agonists or activators of the stimulator of interferon genes (STING) or mitochondrial antiviral signaling protein (MAVS) pathways. After 24 h of incubation, the cytokine profile was analyzed using ELISA and Meso Scale Discovery techniques. RESULTS: ABF suppressed production of IL-6, TNF-α, CXCL10, and IL-10 by PBMCs from SLE patients and HCs following stimulation with specific agonists. ABF also reduced IFN-у production by stimulated PBMCs from HCs. CONCLUSIONS: Our data show that ABF has an immunomodulatory effect on the production of key cytokines in the pathogenesis of SLE. These results suggest that ABF or ABF components hold potential as a novel treatment option for SLE.


Subject(s)
Helminths , Lupus Erythematosus, Systemic , Nucleic Acids , Adult , Animals , Humans , Immunity, Innate , Leukocytes, Mononuclear/metabolism , Nucleic Acids/metabolism
6.
J Immunol ; 204(11): 3042-3055, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32284331

ABSTRACT

Fermentable dietary fibers promote the growth of beneficial bacteria, can enhance mucosal barrier integrity, and reduce chronic inflammation. However, effects on intestinal type 2 immune function remain unclear. In this study, we used the murine whipworm Trichuris muris to investigate the effect of the fermentable fiber inulin on host responses to infection regimes that promote distinct Th1 and Th2 responses in C57BL/6 mice. In uninfected mice, dietary inulin stimulated the growth of beneficial bacteria, such as Bifidobacterium (Actinobacteria) and Akkermansia (Verrucomicrobia). Despite this, inulin prevented worm expulsion in normally resistant mice, instead resulting in chronic infection, whereas mice fed an equivalent amount of nonfermentable fiber (cellulose) expelled worms normally. Lack of expulsion in the mice fed inulin was accompanied by a significantly Th1-skewed immune profile characterized by increased T-bet+ T cells and IFN-γ production in mesenteric lymph nodes, increased expression of Ido1 in the cecum, and a complete absence of mast cell and IgE production. Furthermore, the combination of dietary inulin and high-dose T. muris infection caused marked dysbiosis, with expansion of the Firmicutes and Proteobacteria phyla, near elimination of Bacteroidetes, and marked reductions in cecal short-chain fatty acids. Neutralization of IFN-γ during infection abrogated Ido1 expression and was sufficient to restore IgE production and worm expulsion in inulin-fed mice. Our results indicate that, whereas inulin promoted gut health in otherwise healthy mice, during T. muris infection, it exacerbated inflammatory responses and dysbiosis. Thus, the positive effects of fermentable fiber on gut inflammation appear to be context dependent, revealing a novel interaction between diet and infection.


Subject(s)
Dietary Fiber/metabolism , Inflammation/immunology , Inulin/metabolism , Th1 Cells/immunology , Th2 Cells/immunology , Trichuriasis/immunology , Trichuris/physiology , Animals , Disease Progression , Dysbiosis , Fermentation , Gastrointestinal Microbiome , Host-Pathogen Interactions , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/metabolism , Mice , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
7.
BMC Vet Res ; 17(1): 62, 2021 Jan 29.
Article in English | MEDLINE | ID: mdl-33514383

ABSTRACT

Increasing evidence suggests that nutritional manipulation of the commensal gut microbiota (GM) may play a key role in maintaining animal health and production in an era of reduced antimicrobial usage. Gastrointestinal helminth infections impose a considerable burden on animal performance, and recent studies suggest that infection may substantially alter the composition and function of the GM. Here, we discuss the potential interactions between different bioactive dietary components (prebiotics, probiotics and phytonutrients) and helminth infection on the GM in livestock. A number of recent studies suggest that host diet can strongly influence the nature of the helminth-GM interaction. Nutritional manipulation of the GM may thus impact helminth infection, and conversely infection may also influence how the GM responds to dietary interventions. Moreover, a dynamic interaction exists between helminths, the GM, intestinal immune responses, and inflammation. Deciphering the mechanisms underlying the diet-GM-helminth axis will likely inform future helminth control strategies, as well as having implications for how health-promoting feed additives, such as probiotics, can play a role in sustainable animal production.


Subject(s)
Diet , Gastrointestinal Diseases/veterinary , Gastrointestinal Microbiome/physiology , Helminthiasis, Animal/pathology , Animals , Gastrointestinal Diseases/parasitology , Helminths , Intestinal Diseases, Parasitic , Livestock/microbiology , Livestock/parasitology , Prebiotics , Probiotics
8.
Anal Chem ; 92(15): 10274-10282, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32631050

ABSTRACT

The mechanical properties of extracellular vesicles (EVs) are known to influence their biological function, in terms of, e.g., cellular adhesion, endo/exocytosis, cellular uptake, and mechanosensing. EVs have a characteristic nanomechanical response which can be probed via force spectroscopy (FS) and exploited to single them out from nonvesicular contaminants or to discriminate between subtypes. However, measuring the nanomechanical characteristics of individual EVs via FS is a labor-intensive and time-consuming task, usually limiting this approach to specialists. Herein, we describe a simple atomic force microscopy based experimental procedure for the simultaneous nanomechanical and morphological analysis of several hundred individual nanosized EVs within the hour time scale, using basic AFM equipment and skills and only needing freely available software for data analysis. This procedure yields a "nanomechanical snapshot" of an EV sample which can be used to discriminate between subpopulations of vesicular and nonvesicular objects in the same sample and between populations of vesicles with similar sizes but different mechanical characteristics. We demonstrate the applicability of the proposed approach to EVs obtained from three very different sources (human colorectal carcinoma cell culture, raw bovine milk, and Ascaris suum nematode excretions), recovering size and stiffness distributions of individual vesicles in a sample. EV stiffness values measured with our high-throughput method are in very good quantitative accord with values obtained by FS techniques which measure EVs one at a time. We show how our procedure can detect EV samples contamination by nonvesicular aggregates and how it can quickly attest the presence of EVs even in samples for which no established assays and/or commercial kits are available (e.g., Ascaris EVs), thus making it a valuable tool for the rapid assessment of EV samples during the development of isolation/enrichment protocols by EV researchers. As a side observation, we show that all measured EVs have a strikingly similar stiffness, further reinforcing the hypothesis that their mechanical characteristics could have a functional role.


Subject(s)
Extracellular Vesicles/chemistry , High-Throughput Screening Assays , Microscopy, Atomic Force , Nanotechnology , Animals , Ascaris suum/chemistry , Cattle , HCT116 Cells , Humans , Liposomes/chemistry , Milk/chemistry
9.
J Infect Dis ; 217(2): 310-319, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29136163

ABSTRACT

Ascaris suum is a helminth parasite of pigs closely related to its human counterpart, A. lumbricoides, which infects almost 1 billion people. Ascaris is thought to modulate host immune and inflammatory responses, which may drive immune hyporesponsiveness during chronic infections. Using transcriptomic analysis, we show here that pigs with a chronic A. suum infection have a substantial suppression of inflammatory pathways in the intestinal mucosa, with a broad downregulation of genes encoding cytokines and antigen-processing and costimulatory molecules. A. suum body fluid (ABF) suppressed similar transcriptional pathways in human dendritic cells (DCs) in vitro. DCs exposed to ABF secreted minimal amounts of cytokines and had impaired production of cyclooxygengase-2, altered glucose metabolism, and reduced capacity to induce interferon-gamma production in T cells. Our in vivo and in vitro data provide an insight into mucosal immune modulation during Ascaris infection, and show that A. suum profoundly suppresses immune and inflammatory pathways.


Subject(s)
Ascariasis/pathology , Ascaris suum/immunology , Dendritic Cells/immunology , Immune Tolerance , Intestinal Mucosa/pathology , Animals , Ascariasis/immunology , Cells, Cultured , Disease Models, Animal , Gene Expression Profiling , Humans , Intestinal Mucosa/immunology , Models, Biological , Swine
10.
FASEB J ; 31(2): 719-731, 2017 02.
Article in English | MEDLINE | ID: mdl-27806992

ABSTRACT

Clinical trials have shown that administration of the nematode Trichuris suis can be beneficial in treating various immune disorders. To provide insight into the mechanisms by which this worm suppresses inflammatory responses, an active component was purified from T. suis soluble products (TsSPs) that suppress---- TNF and IL-12 secretion from LPS-activated human dendritic cells (DCs). Analysis by liquid chromatography tandem mass spectrometry identified this compound as prostaglandin (PG)E2. The purified compound showed similar properties compared with TsSPs and commercial PGE2 in modulating LPS-induced expression of many cytokines and chemokines and in modulating Rab7B and P2RX7 expression in human DCs. Furthermore, the TsSP-induced reduction of TNF secretion from DCs is reversed by receptor antagonists for EP2 and EP4, indicating PGE2 action. T. suis secretes extremely high amounts of PGE2 (45-90 ng/mg protein) within their excretory/secretory products but few related lipid mediators as established by metabololipidomic analysis. Culture of T. suis with several cyclooxygenase (COX) inhibitors that inhibit mammalian prostaglandin synthesis affected the worm's motility but did not inhibit PGE2 secretion, suggesting that the worms can synthesize PGE2 via a COX-independent pathway. We conclude that T. suis secretes PGE2 to suppress proinflammatory responses in human DCs, thereby modulating the host's immune response.-Laan, L. C., Williams, A. R., Stavenhagen, K., Giera, M., Kooij, G., Vlasakov, I., Kalay, H., Kringel, H., Nejsum, P., Thamsborg, S. M., Wuhrer, M., Dijkstra, C. D., Cummings, R. D., van Die, I. The whipworm (Trichuris suis) secretes prostaglandin E2 to suppress proinflammatory properties in human dendritic cells.


Subject(s)
Dendritic Cells/metabolism , Dinoprostone/metabolism , Dinoprostone/pharmacology , Inflammation/metabolism , Trichuris/metabolism , Animals , Cells, Cultured , Gene Expression Regulation/drug effects , Humans , Lipopolysaccharides/toxicity , Molecular Structure , Species Specificity
12.
Nature ; 479(7374): 529-33, 2011 Oct 26.
Article in English | MEDLINE | ID: mdl-22031327

ABSTRACT

Parasitic diseases have a devastating, long-term impact on human health, welfare and food production worldwide. More than two billion people are infected with geohelminths, including the roundworms Ascaris (common roundworm), Necator and Ancylostoma (hookworms), and Trichuris (whipworm), mainly in developing or impoverished nations of Asia, Africa and Latin America. In humans, the diseases caused by these parasites result in about 135,000 deaths annually, with a global burden comparable with that of malaria or tuberculosis in disability-adjusted life years. Ascaris alone infects around 1.2 billion people and, in children, causes nutritional deficiency, impaired physical and cognitive development and, in severe cases, death. Ascaris also causes major production losses in pigs owing to reduced growth, failure to thrive and mortality. The Ascaris-swine model makes it possible to study the parasite, its relationship with the host, and ascariasis at the molecular level. To enable such molecular studies, we report the 273 megabase draft genome of Ascaris suum and compare it with other nematode genomes. This genome has low repeat content (4.4%) and encodes about 18,500 protein-coding genes. Notably, the A. suum secretome (about 750 molecules) is rich in peptidases linked to the penetration and degradation of host tissues, and an assemblage of molecules likely to modulate or evade host immune responses. This genome provides a comprehensive resource to the scientific community and underpins the development of new and urgently needed interventions (drugs, vaccines and diagnostic tests) against ascariasis and other nematodiases.


Subject(s)
Ascaris suum/genetics , Genome, Helminth/genetics , Animals , Antinematodal Agents , Ascariasis/drug therapy , Ascariasis/parasitology , Ascaris suum/drug effects , Drug Design , Genes, Helminth/genetics , Genomics , Molecular Sequence Annotation , Molecular Targeted Therapy
13.
Parasitol Res ; 115(3): 1027-37, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26573519

ABSTRACT

Avian schistosomes are widespread parasites of snails and waterfowl and may cause cercarial dermatitis (swimmer's itch) in humans, a disease that is frequently reported in European countries. These parasites are known to occur in Denmark, but here, we applied a new approach using molecular tools to identify the parasites at species level. In order to do that, 499 pulmonate freshwater snails (Radix sp., Lymnaea stagnalis, Stagnicola sp. and Planorbarius corneus) were sampled from 12 lakes, ponds, and marshes in the greater Copenhagen area. Avian schistosome cercariae were identified by microscopy and subjected to molecular investigation by sequencing and phylogenetic analysis of the 5.8S and ITS2 ribosomal DNA for species identification. Additionally, snail hosts belonging to the genus Radix were identified by sequencing and phylogenetic analysis of partial ITS2 ribosomal DNA. Three out of 499 snails shed different species of Trichobilharzia cercariae: Trichobilharzia szidati was isolated from L. stagnalis, Trichobilharzia franki from Radix auricularia and Trichobilharzia regenti from Radix peregra. In the light of the public health risk represented by bird schistosomes, these findings are of concern and, particularly, the presence of the potentially neuro-pathogenic species, T. regenti, in Danish freshwaters calls for attention.


Subject(s)
Dermatitis/parasitology , Schistosomatidae/pathogenicity , Schistosomiasis/parasitology , Skin Diseases, Parasitic/parasitology , Snails/parasitology , Animals , Bird Diseases/parasitology , Birds , Cercaria/classification , Cercaria/genetics , Cercaria/isolation & purification , Cercaria/pathogenicity , DNA, Helminth/chemistry , DNA, Helminth/isolation & purification , DNA, Ribosomal/chemistry , Denmark/epidemiology , Dermatitis/epidemiology , Fresh Water/parasitology , Genetic Variation , Humans , Lymnaea/parasitology , Phylogeny , Schistosomatidae/classification , Schistosomatidae/genetics , Schistosomatidae/isolation & purification , Schistosomiasis/epidemiology , Skin Diseases, Parasitic/epidemiology
14.
Parasitol Res ; 114(7): 2507-15, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25877388

ABSTRACT

This histopathological study was carried out in order to investigate the cellular response in the jejunum to Ascaridia galli during the first 7 weeks of infection. Fourty-two ISA Brown chickens (7 weeks old) were infected orally with 500 embryonated A. galli eggs each while 28 chickens were left as uninfected controls. Six infected and four control chickens were necropsied at each time point 3, 7, 10, 14, 21, 28 and 42 days post-infection (dpi). Samples for histopathology were taken from three sites of the jejunoileum. Significantly higher eosinophil counts were seen in infected chickens compared to uninfected at 3, 7, 10, 14 and 28 dpi (P < 0.01). In both groups, the initial number of mast cells was high, but this high level of mast cells remained for a longer period in the infected group compared to the control group. Significantly higher counts were thus found in the infected group at 21 (P < 0.001), 28 (P < 0.01) and 42 dpi (P < 0.05). A. galli infection induced changes in the mucosal thickness as reduced villi length at 7, 10, 14, 21 and 28 dpi and in the degree of general cellular infiltration in the lamina propria of the mucosal layer. No adult worms were seen during the experiment; therefore, A. galli larvae have elicited a moderate cellular response in the lamina propria, mainly consisting of eosinophils in the early phase and later of mast cells.


Subject(s)
Ascaridia/physiology , Ascaridiasis/veterinary , Jejunum/pathology , Poultry Diseases/pathology , Animals , Ascaridiasis/parasitology , Ascaridiasis/pathology , Chickens , Intestine, Small/parasitology , Intestine, Small/pathology , Jejunum/parasitology , Larva/physiology , Ovum/physiology , Poultry Diseases/parasitology
15.
Trop Anim Health Prod ; 47(8): 1523-30, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26210397

ABSTRACT

Few studies have been carried out in Africa to estimate the prevalence of Taenia hydatigena. With the aim to determine the prevalence of T. hydatigena in slaughtered pigs and small ruminants (goats and sheep) in Mbeya, Tanzania, two cross-sectional surveys were carried out investigating pigs in April to May 2014 and small ruminants in September 2012. In total, 243 pigs were examined post-mortem for T. hydatigena cysts which were found in 16 (6.6 %) pigs. The majority (80 %) of cysts were found on the omentum and the rest on the liver (20 %), all on the visceral surface. Two pigs were also found infected with Taenia solium but showed no signs of other infections. A total of 392 goats and 27 sheep were examined post-mortem, and the prevalence of T. hydatigena was similar in goats and sheep with 45.7 and 51.9 %, respectively. DNA sequencing of the mitochondrial cytochrome c oxidase subunit 1 gene (cox1) from a subsample of metacestodes from goats and sheep confirmed the T. hydatigena infection. The prevalence found in small ruminants was comparable to other studies conducted in Africa, but for pigs, it is one of the highest recorded to date. The present study also confirms the occurrence of T. hydatigena and T. solium in pigs from Mbeya. Further studies are needed to determine the impact of T. hydatigena on production under sub-Saharan conditions and the financial consequences for smallholder farmers.


Subject(s)
Cysticercosis/veterinary , Goat Diseases/parasitology , Goats/parasitology , Sheep Diseases/parasitology , Sheep, Domestic/parasitology , Swine Diseases/parasitology , Taenia solium/isolation & purification , Africa , Animals , Cross-Sectional Studies , Female , Goat Diseases/epidemiology , Male , Phylogeny , Prevalence , Sheep/parasitology , Sheep Diseases/epidemiology , Sus scrofa/parasitology , Swine/parasitology , Swine Diseases/epidemiology , Tanzania/epidemiology
16.
J Infect Dis ; 210(6): 932-41, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24688073

ABSTRACT

BACKGROUND: The roundworm Ascaris lumbricoides infects 0.8 billion people worldwide, and Ascaris suum infects innumerable pigs across the globe. The extent of natural cross-transmission of Ascaris between pig and human hosts in different geographical settings is unknown, warranting investigation. METHODS: Adult Ascaris organisms were obtained from humans and pigs in Europe, Africa, Asia, and Latin America. Barcodes were assigned to 536 parasites on the basis of sequence analysis of the mitochondrial cytochrome c oxidase I gene. Genotyping of 410 worms was also conducted using a panel of microsatellite markers. Phylogenetic, population genetic, and Bayesian assignment methods were used for analysis. RESULTS: There was marked genetic segregation between worms originating from human hosts and those originating from pig hosts. However, human Ascaris infections in Europe were of pig origin, and there was evidence of cross-transmission between humans and pigs in Africa. Significant genetic differentiation exists between parasite populations from different countries, villages, and hosts. CONCLUSIONS: In conducting an analysis of variation within Ascaris populations from pig and human hosts across the globe, we demonstrate that cross-transmission takes place in developing and developed countries, contingent upon epidemiological potential and local phylogeography. Our results provide novel insights into the transmission dynamics and speciation of Ascaris worms from humans and pigs that are of importance for control programs.


Subject(s)
Ascariasis/epidemiology , Molecular Epidemiology , Swine Diseases/epidemiology , Animals , Ascariasis/veterinary , Ascaris/genetics , Cyclooxygenase 1/genetics , DNA, Helminth/genetics , Haplotypes/genetics , Humans , Microsatellite Repeats/genetics , Swine , Swine Diseases/parasitology , Zoonoses/epidemiology , Zoonoses/genetics , Zoonoses/parasitology
17.
Parasitology ; 141(6): 777-87, 2014 May.
Article in English | MEDLINE | ID: mdl-24709292

ABSTRACT

Two single nucleotide polymorphisms (SNP TXNIP and SNP ARNT), both on chromosome 4, have been reported to be associated with roundworm (Ascaris suum) burden in pigs. In the present study, we selected pigs with two SNP TXNIP genotypes (AA; n = 24 and AB; n = 24), trickle-infected them with A. suum from 8 weeks of age until necropsy 8 weeks later, and tested the hypothesis that pigs with the AA genotype would have higher levels of resistance than pigs of AB genotype. We used different indicators of resistance (worm burden, fecal egg counts (FEC), number of liver white spots and A. suum-specific serum IgG antibody levels). Pigs of the AA genotype had lower mean macroscopic worm burden (2.4 vs 19.3; P = 0.06), lower mean total worm burden (26.5 vs 70.1; P = 0.09) and excreted fewer A. suum eggs at week 8 PI (mean number of eggs/g feces: 238 vs 1259; P = 0.14) than pigs of the AB genotype, as expected based on prior associations. The pigs were also genotyped at another locus (SNP ARNT) which showed a similar trend. This study provides suggestive evidence that resistant pigs may be selected using a genetic marker, TXNIP, and provides further support to the quantitative trait locus on chromosome 4.


Subject(s)
Ascariasis/veterinary , Ascaris suum/physiology , Disease Resistance/genetics , Polymorphism, Single Nucleotide/genetics , Swine Diseases/immunology , Alleles , Animals , Ascariasis/immunology , Ascariasis/parasitology , Feces/parasitology , Female , Genetic Markers/genetics , Genotype , Liver/parasitology , Lung/parasitology , Male , Parasite Egg Count , Rabbits , Swine , Swine Diseases/parasitology
18.
NPJ Precis Oncol ; 8(1): 103, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760427

ABSTRACT

Glioblastoma is a highly heterogeneous tumor whose pathophysiological complexities dictate both the diagnosis of disease severity as well as response to therapy. Conventional diagnostic tools and standard treatment regimens have only managed to achieve limited success in the management of patients suspected of glioblastoma. Extracellular vesicles are an emerging liquid biopsy tool that has shown great promise in resolving the limitations presented by the heterogeneous nature of glioblastoma. Here we discuss the contrasting yet interdependent dual role of extracellular vesicles as communication agents that contribute to the progression of glioblastoma by creating a heterogeneous microenvironment and as a liquid biopsy tool providing an opportunity to accurately identify the disease severity and progression.

19.
Front Immunol ; 15: 1328401, 2024.
Article in English | MEDLINE | ID: mdl-38481989

ABSTRACT

Background: Ascaris lumbricoides cystatin (Al-CPI) prevents the development of allergic airway inflammation and dextran-induced colitis in mice models. It has been suggested that helminth-derived cystatins inhibit cathepsins in dendritic cells (DC), but their immunomodulatory mechanisms are unclear. We aimed to analyze the transcriptional profile of human monocyte-derived DC (moDC) upon stimulation with Al-CPI to elucidate target genes and pathways of parasite immunomodulation. Methods: moDC were generated from peripheral blood monocytes from six healthy human donors of Denmark, stimulated with 1 µM of Al-CPI, and cultured for 5 hours at 37°C. RNA was sequenced using TrueSeq RNA libraries and the NextSeq 550 v2.5 (75 cycles) sequencing kit (Illumina, Inc). After QC, reads were aligned to the human GRCh38 genome using Spliced Transcripts Alignment to a Reference (STAR) software. Differential expression was calculated by DESEq2 and expressed in fold changes (FC). Cell surface markers and cytokine production by moDC were evaluated by flow cytometry. Results: Compared to unstimulated cells, Al-CPI stimulated moDC showed differential expression of 444 transcripts (|FC| ≥1.3). The top significant differences were in Kruppel-like factor 10 (KLF10, FC 3.3, PBH = 3 x 10-136), palladin (FC 2, PBH = 3 x 10-41), and the low-density lipoprotein receptor (LDLR, FC 2.6, PBH = 5 x 10-41). Upregulated genes were enriched in regulation of cholesterol biosynthesis by sterol regulatory element-binding proteins (SREBP) signaling pathways and immune pathways. Several genes in the cholesterol biosynthetic pathway showed significantly increased expression upon Al-CPI stimulation, even in the presence of lipopolysaccharide (LPS). Regarding the pathway of negative regulation of immune response, we found a significant decrease in the cell surface expression of CD86, HLA-DR, and PD-L1 upon stimulation with 1 µM Al-CPI. Conclusion: Al-CPI modifies the transcriptome of moDC, increasing several transcripts encoding enzymes involved in cholesterol biosynthesis and SREBP signaling. Moreover, Al-CPI target several transcripts in the TNF-alpha signaling pathway influencing cytokine release by moDC. In addition, mRNA levels of genes encoding KLF10 and other members of the TGF beta and the IL-10 families were also modified by Al-CPI stimulation. The regulation of the mevalonate pathway and cholesterol biosynthesis suggests new mechanisms involved in DC responses to helminth immunomodulatory molecules.


Subject(s)
Cystatins , Monocytes , Humans , Animals , Mice , Ascaris lumbricoides , Mevalonic Acid/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Cell Differentiation , Cytokines/metabolism , Inflammation/metabolism , Immunity , Dendritic Cells , RNA/metabolism
20.
Parasit Vectors ; 17(1): 225, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755679

ABSTRACT

BACKGROUND: The giant roundworm Ascaris is an intestinal nematode, causing ascariasis by infecting humans and pigs worldwide. Recent estimates suggest that Ascaris infects over half a billion people, with chronic infections leading to reduced growth and cognitive ability. Ascariasis affects innumerable pigs worldwide and is known to reduce production yields via decreased growth and condemnation of livers. The predominant anthelminthic drugs used to treat ascariasis are the benzimidazoles. Benzimidazoles interact with ß-tubulins and block their function, and several benzimidazole resistance-associated mutations have been described in the ß-tubulins of ruminant nematodes. Recent research on ascarids has shown that these canonical benzimidazole resistance-associated mutations are likely not present in the ß-tubulins of Ascaris, Ascaridia or Parascaris, even in phenotypically resistant populations. METHODS: To further determine the putative absence of key ß-tubulin polymorphisms, we screened two ß-tubulin isotypes of Ascaris, highly expressed in adult worms. Using adult and egg samples of Ascaris obtained from pigs and humans worldwide, we performed deep amplicon sequencing to look for canonical resistance-associated mutations in Ascaris ß-tubulins. Subsequently, we examined these data in closer detail to study the population dynamics of Ascaris and genetic diversity within the two isotypes and tested whether genotypes appeared to partition across human and pig hosts. RESULTS: In the 187 isolates, 69 genotypes were found, made up of eight haplotypes of ß-tubulin isotype A and 20 haplotypes of isotype B. Single nucleotide polymorphisms were seen at 14 and 37 positions for ß-tubulin isotype A and isotype B, respectively. No evidence of any canonical benzimidazole resistance-associated mutations was found in either human- or pig-derived Ascaris isolates. There was, however, a difference in the genetic diversity of each isotype and distribution of ß-tubulin genotypes between human- and pig-derived Ascaris. Statistical tests of population differentiation show significant differences (p < 0.001) between pig- and human-derived worms; however, more diversity was seen between worms from different populations than worms from different hosts. CONCLUSIONS: Our work suggests an absence of canonical ß-tubulin mutations within Ascaris, but alternative modes of anthelminthic resistance may emerge necessitating continued genetic scrutiny alongside monitoring of drug efficacy.


Subject(s)
Anthelmintics , Ascariasis , Ascaris , Benzimidazoles , Drug Resistance , Mutation , Tubulin , Tubulin/genetics , Animals , Benzimidazoles/pharmacology , Drug Resistance/genetics , Ascariasis/parasitology , Ascariasis/veterinary , Ascariasis/drug therapy , Anthelmintics/pharmacology , Swine , Ascaris/genetics , Ascaris/drug effects , Humans , Swine Diseases/parasitology , Swine Diseases/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL